Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Phosphatidylserine receptor BAI1 and apoptotic cells as new promoters of myoblast fusion

Subjects

Abstract

Skeletal muscle arises from the fusion of precursor myoblasts into multinucleated myofibres1,2. Although conserved transcription factors and signalling proteins involved in myogenesis have been identified, upstream regulators are less well understood. Here we report an unexpected discovery that the membrane protein BAI1, previously linked to recognition of apoptotic cells by phagocytes3, promotes myoblast fusion. Endogenous BAI1 expression increased during myoblast fusion, and BAI1 overexpression enhanced myoblast fusion by means of signalling through ELMO/Dock180/Rac1 proteins4. During myoblast fusion, a fraction of myoblasts within the population underwent apoptosis and exposed phosphatidylserine, an established ligand for BAI1 (ref. 3). Blocking apoptosis potently impaired myoblast fusion, and adding back apoptotic myoblasts restored fusion. Furthermore, primary human myoblasts could be induced to form myotubes by adding apoptotic myoblasts, even under normal growth conditions. Mechanistically, apoptotic cells did not directly fuse with the healthy myoblasts, rather the apoptotic cells induced a contact-dependent signalling with neighbours to promote fusion among the healthy myoblasts. In vivo, myofibres from Bai1−/− mice are smaller than those from wild-type littermates. Muscle regeneration after injury was also impaired in Bai1−/− mice, highlighting a role for BAI1 in mammalian myogenesis. Collectively, these data identify apoptotic cells as a new type of cue that induces signalling via the phosphatidylserine receptor BAI1 to promote fusion of healthy myoblasts, with important implications for muscle development and repair.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The phosphatidylserine receptor BAI1 promotes myoblast fusion.
Figure 2: Apoptosis of myoblasts is necessary cue for fusion.
Figure 3: Cell–cell contact through apoptotic myoblasts promotes healthy myoblast fusion.
Figure 4: Defective development of myofibres in Bai1−/− mice.

Similar content being viewed by others

References

  1. Abmayr, S. M. & Pavlath, G. K. Myoblast fusion: lessons from flies and mice. Development 139, 641–656 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chen, E. H. & Olson, E. N. Towards a molecular pathway for myoblast fusion in Drosophila . Trends Cell Biol. 14, 452–460 (2004)

    Article  CAS  PubMed  Google Scholar 

  3. Park, D. et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450, 430–434 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  4. Gumienny, T. L. et al. CED-12/ELMO, a novel member of the CrkII/Dock180/Rac pathway, is required for phagocytosis and cell migration. Cell 107, 27–41 (2001)

    Article  CAS  PubMed  Google Scholar 

  5. Hasegawa, H. et al. DOCK180, a major CRK-binding protein, alters cell morphology upon translocation to the cell membrane. Mol. Cell. Biol. 16, 1770–1776 (1996)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Brugnera, E. et al. Unconventional Rac-GEF activity is mediated through the Dock180-ELMO complex. Nature Cell Biol. 4, 574–582. (2002)

  7. Erickson, M. R., Galletta, B. J. & Abmayr, S. M. Drosophila myoblast city encodes a conserved protein that is essential for myoblast fusion, dorsal closure, and cytoskeletal organization. J. Cell Biol. 138, 589–603 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hakeda-Suzuki, S. et al. Rac function and regulation during Drosophila development. Nature 416, 438–442 (2002)

    Article  CAS  ADS  PubMed  Google Scholar 

  9. Moore, C. A., Parkin, C. A., Bidet, Y. & Ingham, P. W. A role for the Myoblast city homologues Dock1 and Dock5 and the adaptor proteins Crk and Crk-like in zebrafish myoblast fusion. Development 134, 3145–3153 (2007)

    Article  CAS  PubMed  Google Scholar 

  10. Laurin, M. et al. The atypical Rac activator Dock180 (Dock1) regulates myoblast fusion in vivo . Proc. Natl Acad. Sci. USA 105, 15446–15451 (2008)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  11. Vasyutina, E., Martarelli, B., Brakebusch, C., Wende, H. & Birchmeier, C. The small G-proteins Rac1 and Cdc42 are essential for myoblast fusion in the mouse. Proc. Natl Acad. Sci. USA 106, 8935–8940 (2009)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  12. Geisbrecht, E. R. et al. Drosophila ELMO/CED-12 interacts with Myoblast city to direct myoblast fusion and ommatidial organization. Dev. Biol. 314, 137–149 (2008)

    Article  CAS  PubMed  Google Scholar 

  13. Yaffe, D. & Saxel, O. Serial passaging and differentiation of myogenic cells isolated from dystrophic mouse muscle. Nature 270, 725–727 (1977)

    Article  CAS  ADS  PubMed  Google Scholar 

  14. Cornelison, D. D. Context matters: in vivo and in vitro influences on muscle satellite cell activity. J. Cell. Biochem. 105, 663–669 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Pajcini, K. V., Pomerantz, J. H., Alkan, O., Doyonnas, R. & Blau, H. M. Myoblasts and macrophages share molecular components that contribute to cell-cell fusion. J. Cell Biol. 180, 1005–1019 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shutes, A. et al. Specificity and mechanism of action of EHT 1864, a novel small molecule inhibitor of Rac family small GTPases. J. Biol. Chem. 282, 35666–35678 (2007)

    Article  CAS  PubMed  Google Scholar 

  17. van den Eijnde, S. M. et al. Transient expression of phosphatidylserine at cell-cell contact areas is required for myotube formation. J. Cell Sci. 114, 3631–3642 (2001)

    CAS  PubMed  Google Scholar 

  18. Jeong, J. & Conboy, I. M. Phosphatidylserine directly and positively regulates fusion of myoblasts into myotubes. Biochem. Biophys. Res. Commun. 414, 9–13 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chekeni, F. B. et al. Pannexin 1 channels mediate 'find-me' signal release and membrane permeability during apoptosis. Nature 467, 863–867 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  20. Hasty, P. et al. Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin gene. Nature 364, 501–506 (1993)

    Article  CAS  ADS  PubMed  Google Scholar 

  21. Kamentsky, L. et al. Improved structure, function and compatibility for CellProfiler: modular high-throughput image analysis software. Bioinformatics 27, 1179–1180 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yan, Z. et al. Highly coordinated gene regulation in mouse skeletal muscle regeneration. J. Biol. Chem. 278, 8826–8836 (2003)

    Article  CAS  PubMed  Google Scholar 

  23. Grounds, M. D., Radley, H. G., Lynch, G. S., Nagaraju, K. & De Luca, A. Towards developing standard operating procedures for pre-clinical testing in the mdx mouse model of Duchenne muscular dystrophy. Neurobiol. Dis. 31, 1–19 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen, Y. W., Zhao, P., Borup, R. & Hoffman, E. P. Expression profiling in the muscular dystrophies: identification of novel aspects of molecular pathophysiology. J. Cell Biol. 151, 1321–1336 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Haslett, J. N. et al. Gene expression comparison of biopsies from Duchenne muscular dystrophy (DMD) and normal skeletal muscle. Proc. Natl Acad. Sci. USA 99, 15000–15005 (2002)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  26. Bakay, M. et al. Nuclear envelope dystrophies show a transcriptional fingerprint suggesting disruption of Rb-MyoD pathways in muscle regeneration. Brain 129, 996–1013 (2006)

    Article  PubMed  Google Scholar 

  27. Bialek, P. et al. Distinct protein degradation profiles are induced by different disuse models of skeletal muscle atrophy. Physiol. Genomics 43, 1075–1086 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fernando, P., Kelly, J. F., . Balazsi, K., Slack, R. S. & Megeney, L. A. Caspase 3 activity is required for skeletal muscle differentiation. Proc. Natl Acad. Sci. USA 99, 11025–11030 (2002)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  29. Phaneuf, S. & Leeuwenburgh, C. Apoptosis and exercise. Med. Sci. Sports Exerc. 33, 393–396 (2001)

    Article  CAS  PubMed  Google Scholar 

  30. Ravichandran, K. S. Find-me and eat-me signals in apoptotic cell clearance: progress and conundrums. J. Exp. Med. 207, 1807–1817 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sokolowski, J. D. et al. Brain-specific angiogenesis inhibitor-1 expression in astrocytes and neurons: implications for its dual function as an apoptotic engulfment receptor. Brain Behav. Immun. 25, 915–921 (2011)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Ravichandran laboratory for their valuable suggestions at many stages of this work. We also thank L. Haney, A. Bruce and A. Dutta for technical suggestions and assistance and M. Hufford and A. Fond for help with statistical analysis. We thank members of the University of Virginia Flow Cytometry Core, Research Histology Core, and Gene Targeting and Transgenic Facility for cell sorting, histological services and transgenic mouse generation. This work was supported by a grant from the National Institute of General Medical Sciences/National Institutes of Health and the Center for Cell Clearance at the University of Virginia.

Author information

Authors and Affiliations

Authors

Contributions

A.E.H.-H. designed, performed and analysed most of the experiments in this study with input from K.S.R. C.S.L. generated and supplied the Bai1−/− mice, and provided GST-tagged BAI1 TSR. J.M.K. assisted with time-lapse and shRNA studies, and myofibre cross-sectional area analyses. S.A. helped with the in vivo muscle regeneration and ex vivo primary myoblast cultures. J.D.S. processed, stained and analysed the mouse embryonic tissues. A.L.K. provided phosphatidylserine liposomes for these studies. J.A.C. and Z.Y. assisted with the cardiotoxin injury model. J.W.M. provided intellectual input on the in vivo BAI1 studies. A.E.H.-H. and K.S.R. wrote the manuscript with comments from co-authors.

Corresponding author

Correspondence to Kodi S. Ravichandran.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-7. (PDF 39261 kb)

A population of myoblasts undergo cell death during fusion and remain in close contact with nascent myotubes

This video shows C2C12 fusing myoblasts labeled with TO-PRO-3 (red) to identify apoptotic cells. The time-lapse covers a period of 72 hours during which time a number of myoblasts die but do not appear to be phagocytosed. These TO-PRO-3 positive cells are passed along the newly forming myotubes culture maintain contact with the myotubes for the duration of the video. (MOV 28781 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hochreiter-Hufford, A., Lee, C., Kinchen, J. et al. Phosphatidylserine receptor BAI1 and apoptotic cells as new promoters of myoblast fusion. Nature 497, 263–267 (2013). https://doi.org/10.1038/nature12135

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12135

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing